PPARG Human (1-477)

Peroxisome Proliferator Activated Receptor Gamma Human Recombinant, (1-477 a.a)
Shipped with Ice Packs
In Stock

Description

Functional Roles in Physiology and Disease

PPARG Human (1-477) retains the biological activities of endogenous PPARγ1:

Metabolic Regulation

  • Adipogenesis: Directs adipocyte differentiation via target genes like aP2 and GyK .

  • Glucose homeostasis: Enhances insulin sensitivity by promoting GLUT4 translocation .

  • Lipid metabolism: Regulates fatty acid uptake and storage through CD36 and LPL .

Immune Modulation

  • Suppresses pro-inflammatory cytokines (e.g., TNF-α, IL-6) by antagonizing NF-κB .

  • Promotes anti-inflammatory macrophage polarization and Treg differentiation .

Cancer and Disease Links

  • Oncogenic potential: PPARγ fusion proteins (e.g., PAX8-PPARγ) drive thyroid carcinoma .

  • Hypertension: Loss-of-function mutations correlate with elevated blood pressure .

Ligand Binding and Activation

  • The LBD accommodates diverse ligands, including polyunsaturated fatty acids and synthetic TZDs, stabilizing helix 12 for co-activator recruitment .

  • Phosphorylation at Ser112 (via MEK/ERK) reduces transcriptional activity, contributing to insulin resistance .

Pharmacological Insights

  • Partial agonists (e.g., INT131) retain metabolic benefits without TZD-associated side effects .

  • The Pro12Ala polymorphism (rs1801282) reduces T2DM risk by 18–25% in European populations .

Applications in Biomedical Research

PPARG Human (1-477) is utilized in:

  • Drug screening: Assess ligand binding via TR-FRET or fluorescence polarization assays .

  • Protein interaction studies: Identifies co-activators (e.g., PGC-1α) and co-repressors (e.g., NCoR) .

  • Functional assays: Measures transcriptional activity in reporter gene systems .

Table 2: Experimental Use Cases

ApplicationMethodKey FindingsCitation
Ligand bindingX-ray crystallographyTZDs stabilize AF-2 via helix 12
Co-regulatorMammalian two-hybrid assayNCoR interaction requires Y477 residue
Disease modelingCellular reporter assaysPPARγΔ5 isoforms impair adipogenesis

Clinical and Therapeutic Relevance

  • T2DM treatment: Rosiglitazone (a TZD) improves insulin sensitivity but increases cardiovascular risks .

  • Cancer therapy: PPARγ agonists inhibit proliferation in breast and prostate cancer cells .

  • Inflammatory diseases: PPARγ activation alleviates atherosclerosis and autoimmune disorders .

Product Specs

Introduction
Peroxisome proliferators, classified as non-genotoxic carcinogens, are believed to influence cellular processes by interacting with a group of nuclear hormone receptors known as peroxisome proliferator-activated receptors (PPARs). These receptors function by binding to specific DNA sequences upon activation by their respective ligands, thereby stimulating the transcription of target genes. Research suggests that PPARs can be activated by peroxisome proliferators, including clofibric acid, nafenopin, and WY-14,643, as well as certain fatty acids.
Description
This product consists of the PPARG protein (amino acids 1-477) from humans, recombinantly expressed in E. coli. It is fused to a GST tag for detection and purification purposes. The protein is purified using proprietary chromatographic methods, ensuring high purity and antibody reactivity.
Physical Appearance
The product is a clear solution that has been sterilized by filtration.
Formulation
The PPARG protein is supplied at a concentration of 0.1 mg/ml in a buffer solution containing 50 mM Tris-HCl and 10 mM reduced L-glutathione.
Applications
This product is suitable for use in various applications, including enzyme-linked immunosorbent assays (ELISA), inhibition assays, and Western blotting.
Stability
To maintain product stability, store the vial at a temperature between -20°C and -80°C. Under these conditions, the protein remains stable for a period of 12 months. Repeated freezing and thawing of the product should be avoided.
Synonyms
Peroxisome proliferator-activated receptor gamma, PPAR-gamma, PPARG, NR1C3, PPARG1, PPARG2.
Source
Escherichia Coli.

Q&A

What is PPARG and what are its primary functions in human biology?

PPARG (NR1C3) is a member of the nuclear receptor superfamily that functions as a ligand-activated transcription factor. It was identified following the discovery of PPARα (NR1C1) in 1990 and was subsequently cloned in Xenopus and humans . PPARG primarily regulates glucose and lipid metabolism, adipocyte differentiation, and controls inflammatory responses. Methodologically, researchers should approach PPARG studies through integrated analyses of its genomic binding patterns (ChIP-seq), transcriptional outputs (RNA-seq), and metabolic effects in relevant cell types and tissues.

What are the key structural domains of PPARG Human (1-477) and their functions?

PPARG Human (1-477) encompasses the functional domains of this nuclear receptor, including the N-terminal domain, DNA-binding domain, hinge region, and ligand-binding domain. The ligand-binding domain contains the activation function-2 (AF-2) that undergoes conformational changes upon ligand binding, facilitating interactions with co-activators such as PGC-1α and E1A binding protein p300 (EP300) . Researchers can utilize techniques like hydrogen/deuterium exchange mass spectrometry and X-ray crystallography to study domain-specific structural changes associated with different ligands and co-regulator interactions.

How do the different PPARG isoforms differ in expression and function?

PPARG exists in two main isoforms: PPARγ1 and PPARγ2. PPARγ2 contains an additional 30 amino acids at its N-terminus compared to PPARγ1 . While PPARγ1 is expressed in many tissues including leukocytes and endothelial cells, PPARγ2 expression is normally restricted to adipose tissue but can be induced elsewhere under specific conditions . When designing tissue-specific studies, researchers should account for these differential expression patterns by utilizing isoform-specific PCR primers, antibodies, and appropriate cellular models.

What are the endogenous ligands of PPARG and how do they compare to synthetic agonists?

Several endogenous compounds have been identified as potential PPARG ligands:

  • 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) and related metabolites can activate PPARG, though at concentrations above physiological levels

  • Unsaturated fatty acids such as eicosapentanoic acid, linolenic acids, and linoleic acid (K𝐷 values of 2–50 μM)

  • Oxidized low-density lipoprotein and its components (9-HODE and 13-HODE)

  • Nitrated fatty acids, particularly nitro-linoleic acid (K𝑖 value of 133 nM, comparable to rosiglitazone at 53 nM)

Methodologically, competitive binding assays, reporter gene assays, and thermal shift assays should be employed to comparatively evaluate endogenous versus synthetic ligand potency, efficacy, and selectivity.

How does nitric oxide (NO) signaling intersect with PPARG function?

NO can modify unsaturated fatty acids through nitration, creating potent PPARG ligands. Nitro-linoleic acid has a binding affinity (K𝑖 = 133 nM) that rivals synthetic agonists like rosiglitazone (K𝑖 = 53 nM) and is much higher than unmodified linoleic acid (K𝑖 > 1 μM) . These nitrated fatty acids can covalently bind to PPARG at C285 via Michael addition, activating PPARG genomic signaling . Nitrated fatty acids represent one of the largest pools of active NO derivatives in human plasma, potentially reaching concentrations >1 μM . Researchers investigating this crosslink should implement mass spectrometry techniques to detect nitrated fatty acids in biological samples and employ site-directed mutagenesis of C285 to confirm the mechanistic importance of this residue.

How do thiazolidinediones (TZDs) activate PPARG, and what parallel signaling pathways are involved?

TZDs directly bind to PPARG's ligand-binding domain but also activate parallel signaling pathways:

  • TZDs activate p38 MAPK independent of PPARG in various cell types

  • In endothelial cells, rosiglitazone (RGZ) activation of GPR40 is essential for optimal PPARG genomic signaling

  • RGZ/GPR40/p38 MAPK signaling induces and activates PGC-1α and recruits EP300 to PPARG target genes

What is the significance of the Pro12Ala polymorphism in PPARG research?

The Pro12Ala variant (rs1801282) is one of the most studied PPARG polymorphisms. Research shows variable associations with Type 2 Diabetes Mellitus (T2DM) risk across populations:

  • Protective effects against T2DM have been observed in Japanese, Korean, Greater Middle Eastern, and some European ancestries

  • Increased T2DM risk has been reported in Russian, South Asian (Kashmiri), and mixed ancestry South African populations

These contradictory findings highlight the importance of population-specific effects. Researchers should employ robust methodologies including adequate sample sizes, appropriate controls for population stratification, and consistency in phenotype definitions when studying this variant.

What methodological approaches are recommended for studying PPARG genetic associations?

When investigating PPARG genetic associations, researchers should implement the following methodological framework:

  • Study design considerations:

    • Case-control studies with adequate sample sizes

    • Clear inclusion/exclusion criteria

    • Quality assessment using standardized tools like the Newcastle-Ottawa Scale (NOS)

  • Statistical analysis recommendations:

    • Examine multiple genetic models: allele (G vs. C), homozygote (GG vs. CC), heterozygote (CG vs. CC), additive (GG vs. CG), dominant (GG+CG vs. CC), recessive (GG vs. CC+CG), and co-dominant (CG vs. CC+GG)

    • Test for Hardy-Weinberg equilibrium in control populations

    • Assess heterogeneity using the Cochrane Q-test and I-square index

    • Apply appropriate statistical models based on heterogeneity (random effects model if I² > 50%)

  • Subgroup analysis strategies:

    • Stratify by ancestry categories

    • Consider BMI and age as potential effect modifiers

    • Analyze by publication period to identify temporal trends

How can contradictory findings in PPARG genetic association studies be reconciled?

Contradictory findings in PPARG genetic studies can be reconciled through systematic approaches:

  • Meta-analysis techniques:

    • Pool data from multiple studies to increase statistical power

    • Apply fixed or random effects models based on heterogeneity assessment

    • Calculate pooled odds ratios with confidence intervals

  • Sensitivity analysis methods:

    • Remove studies that don't meet Hardy-Weinberg equilibrium

    • Exclude poor quality studies (NOS score ≤3)

    • Perform leave-one-out analysis to identify influential studies

  • Publication bias assessment:

    • Use Egger's linear regression test

    • Create and analyze funnel plots for asymmetry

    • Apply Duval and Tweedie trim-and-fill technique when publication bias is detected

Researchers should recognize that genetic effects may be context-dependent, varying with ethnicity, environmental factors, and gene-gene interactions.

What post-translational modifications regulate PPARG activity?

PPARG undergoes several post-translational modifications that significantly impact its function:

  • Phosphorylation:

    • ERK1/2 can phosphorylate PPARG, leading to its inactivation and potential contribution to insulin resistance and inflammation

    • p38 MAPK phosphorylates PPARG co-activators including PGC-1α and EP300, enhancing transcriptional activity

  • Sumoylation:

    • PPARG2 can be sumoylated at K395, which is involved in its transrepression activity

    • This modification enables PPARG to tether with nuclear receptor co-repressor and histone deacetylase to NFκB and AP-1 complexes

  • Other modifications:

    • Nitrated and oxidized fatty acids can covalently bind to PPARG at C285 via Michael addition

Researchers studying these modifications should employ mass spectrometry, site-directed mutagenesis, and modification-specific antibodies, along with functional assays to determine their impact on transcriptional activity.

How does p38 MAPK signaling influence PPARG transcriptional activity?

p38 MAPK plays a crucial role in PPARG-mediated transcription through multiple mechanisms:

  • p38 MAPK has been linked to PPARG-dependent adipogenesis in various cell types

  • In brown adipose tissue, p38 MAPK activates PGC-1α and induces expression of PPARG target genes

  • p38 MAPK directly phosphorylates:

    • PGC-1α, a key PPARG co-activator

    • EP300, which facilitates co-activator recruitment to PPARG target genes

  • TZDs can activate p38 MAPK independent of PPARG in multiple cell types, suggesting this pathway contributes to their therapeutic effects

When designing experiments to study this cross-talk, researchers should use specific p38 MAPK inhibitors (e.g., SB203580), phospho-specific antibodies, and phosphorylation-deficient mutants of PGC-1α and EP300 to dissect the precise contributions of this pathway.

What in vitro systems are most appropriate for studying PPARG function?

Researchers investigating PPARG should select experimental systems based on specific research questions:

  • For adipogenic studies:

    • 3T3-L1 preadipocytes

    • Primary human or mouse preadipocytes

    • Mesenchymal stem cells capable of adipogenic differentiation

  • For metabolic regulation:

    • Hepatocytes (HepG2, primary hepatocytes)

    • Skeletal muscle cells (C2C12, primary myotubes)

    • Pancreatic β-cells (INS-1, MIN6)

  • For inflammatory regulation:

    • Macrophages (RAW264.7, THP-1, primary macrophages)

    • Endothelial cells (HUVECs, HAECs)

Experimental readouts should include:

  • PPARG binding assays (competitive binding, thermal shift)

  • Transcriptional activation (luciferase reporters with PPRE elements)

  • Target gene expression (RT-qPCR, RNA-seq)

  • Protein-protein interactions (co-immunoprecipitation, mammalian two-hybrid)

  • Functional outcomes (lipid accumulation, glucose uptake, cytokine production)

What are the key considerations when designing animal models for PPARG research?

When designing animal models for PPARG research, consider:

  • Model selection:

    • Global PPARG knockout is embryonically lethal

    • Tissue-specific conditional knockouts using Cre-lox technology

    • Humanized PPARG models to study human-specific variants

  • Experimental design factors:

    • Age of animals (PPARG function changes with development and aging)

    • Sex differences (metabolic phenotypes often show sexual dimorphism)

    • Dietary conditions (standard chow vs. high-fat diet)

    • Duration of intervention (acute vs. chronic)

  • Phenotypic analyses:

    • Metabolic parameters (glucose tolerance, insulin sensitivity)

    • Tissue histology (adipose, liver, muscle)

    • Molecular readouts (target gene expression, chromatin occupancy)

    • Inflammatory markers (tissue inflammation, systemic cytokines)

Careful consideration of these factors will ensure that animal models appropriately address the specific research question while maintaining translational relevance.

What are the therapeutic applications of PPARG modulation beyond type 2 diabetes?

PPARG modulation shows therapeutic potential in multiple conditions beyond T2DM:

  • Cardiovascular diseases:

    • Endothelial PPARG disruption accelerates diet-induced atherosclerosis

    • PPARG ligands can reduce atherosclerotic lesions in apoE knockout mice

    • Heterozygous mutations (V290M, P467L) in PPARG are linked to hypertension

  • Pulmonary conditions:

    • PPARG deletion in arterial smooth muscle cells leads to pulmonary arterial hypertension

    • PPARG activation protects against acute respiratory distress syndrome

    • PPARG ligands show benefits in murine models of pulmonary hypertension

  • Inflammatory disorders:

    • In endotoxin challenge models, PPARG ligands reduce mortality and organ injury

    • Nitro-oleic acid (PPARG activator) attenuates colitis in experimental inflammatory bowel disease

    • PPARG activation decreases inflammation in models of acute lung injury

Researchers investigating these applications should employ disease-specific models and assess both PPARγ-dependent and independent effects of candidate compounds.

What strategies are being explored to develop improved PPARG modulators with fewer side effects?

Several approaches are being pursued to develop PPARG modulators with improved safety profiles:

  • Targeting specific PPARG conformations:

    • Partial agonists that induce distinct conformational changes

    • Non-agonist PPARG ligands that block phosphorylation without full activation

  • Dual-pathway modulators:

    • Compounds targeting both PPARG and complementary pathways

    • Biased GPR40 agonists that activate p38 MAPK and PPARG without activating ERK1/2

  • Tissue-specific approaches:

    • Compounds with selective tissue distribution

    • Brain-sparing PPARG modulators to avoid weight gain effects

  • Endogenous ligand mimetics:

    • Nitrated fatty acid derivatives that activate PPARG without causing weight gain

    • Modified nitro-oleic acid with improved pharmacokinetic properties

In developing and evaluating these compounds, researchers should implement comprehensive safety assessment including adipogenic potential, fluid retention, bone density effects, and cardiovascular outcomes.

What are the emerging areas of PPARG research with potential for breakthrough discoveries?

Emerging research areas with breakthrough potential include:

  • PPARG in tissue-specific metabolism:

    • Role in brown/beige adipose tissue thermogenesis

    • Regulation of the gut-liver axis in metabolic disease

    • Contributions to brain regulation of energy homeostasis

  • Integrative 'omics approaches:

    • Combined analysis of PPARG cistrome, transcriptome, and metabolome

    • Single-cell resolution of PPARG activity in heterogeneous tissues

    • Spatial transcriptomics to map PPARG activity in intact tissues

  • PPARG in emerging disease contexts:

    • Contributions to NAFLD/NASH pathogenesis and treatment

    • Role in cancer metabolism and inflammation

    • Involvement in neurodegenerative processes

These emerging areas require interdisciplinary approaches combining molecular biology, genetic engineering, advanced imaging, and computational biology.

How can contradictory data on PPARG function be reconciled through improved experimental design?

Researchers can reconcile contradictory findings through several methodological improvements:

  • Context-specific analysis:

    • Define precise experimental conditions (cell type, differentiation state, metabolic status)

    • Consider temporal aspects of PPARG activation (acute vs. chronic)

    • Account for species differences in PPARG biology

  • Multi-level validation:

    • Confirm findings across different experimental systems

    • Employ complementary methodologies to address the same question

    • Validate key findings in human samples whenever possible

  • Mechanistic resolution:

    • Distinguish genomic from non-genomic PPARG effects

    • Separate ligand-dependent from ligand-independent functions

    • Account for post-translational modifications and cofactor availability

  • Quantitative approaches:

    • Apply dose-response relationships rather than single-point measurements

    • Use time-course experiments to capture dynamic responses

    • Employ mathematical modeling to integrate complex datasets

Product Science Overview

Introduction

Peroxisome Proliferator-Activated Receptor Gamma (PPAR-γ or PPARG) is a type II nuclear receptor that functions as a transcription factor. It is encoded by the PPARG gene in humans and plays a crucial role in regulating gene expression related to glucose and lipid metabolism, adipogenesis, and inflammation .

Structure and Function

PPAR-γ is a ligand-activated nuclear receptor that heterodimerizes with the retinoid X receptor (RXR) to regulate the transcription of target genes. The human recombinant form of PPAR-γ, specifically the (1-477 a.a) variant, includes the full-length protein, which is essential for its functional activity .

Biological Role

PPAR-γ is predominantly expressed in adipose tissue, but it is also found in other tissues such as the liver, skeletal muscle, and immune cells. It is a key regulator of adipocyte differentiation and lipid storage. Activation of PPAR-γ by its ligands leads to the transcription of genes involved in fatty acid storage and glucose metabolism, making it a critical player in maintaining energy balance and metabolic homeostasis .

Clinical Significance

PPAR-γ has been extensively studied for its role in metabolic diseases such as diabetes and obesity. Synthetic agonists of PPAR-γ, such as thiazolidinediones (TZDs), are used as therapeutic agents to improve insulin sensitivity in patients with type 2 diabetes. However, the use of these agonists is limited due to associated side effects, including weight gain and cardiovascular risks .

Research and Applications

The recombinant form of PPAR-γ (1-477 a.a) is widely used in research to study its function and regulation. It is also utilized in drug discovery and development to screen for potential PPAR-γ agonists and antagonists. Understanding the molecular mechanisms of PPAR-γ action can lead to the development of more effective and safer therapeutic agents for metabolic disorders .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.